Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 76(15): 4504-15, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27312529

ABSTRACT

Although recent clinical trials of BRAF inhibitor combinations have demonstrated improved efficacy in BRAF-mutant colorectal cancer, emergence of acquired resistance limits clinical benefit. Here, we undertook a comprehensive effort to define mechanisms underlying drug resistance with the goal of guiding development of therapeutic strategies to overcome this limitation. We generated a broad panel of BRAF-mutant resistant cell line models across seven different clinically relevant drug combinations. Combinatorial drug treatments were able to abrogate ERK1/2 phosphorylation in parental-sensitive cells, but not in their resistant counterparts, indicating that resistant cells escaped drug treatments through one or more mechanisms leading to biochemical reactivation of the MAPK signaling pathway. Genotyping of resistant cells identified gene amplification of EGFR, KRAS, and mutant BRAF, as well as acquired mutations in KRAS, EGFR, and MAP2K1 These mechanisms were clinically relevant, as we identified emergence of a KRAS G12C mutation and increase of mutant BRAF V600E allele frequency in the circulating tumor DNA of a patient at relapse from combined treatment with BRAF and MEK inhibitors. To identify therapeutic combinations capable of overcoming drug resistance, we performed a systematic assessment of candidate therapies across the panel of resistant cell lines. Independent of the molecular alteration acquired upon drug pressure, most resistant cells retained sensitivity to vertical MAPK pathway suppression when combinations of ERK, BRAF, and EGFR inhibitors were applied. These therapeutic combinations represent promising strategies for future clinical trials in BRAF-mutant colorectal cancer. Cancer Res; 76(15); 4504-15. ©2016 AACR.


Subject(s)
Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Dosage/genetics , Proto-Oncogene Proteins B-raf/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Gene Amplification , Humans , Signal Transduction
2.
Cancer Discov ; 5(12): 1271-81, 2015 12.
Article in English | MEDLINE | ID: mdl-26432108

ABSTRACT

UNLABELLED: MET inhibition is effective in some patients with MET-amplified esophagogastric cancer (EGC), but understanding acquired and de novo resistance mechanisms will be critical to improving therapy. We identified KRAS mutation as a novel cause of acquired resistance in a patient after a 2-year response to a MET inhibitor. We also observed that 40% to 50% of patients with MET-amplified EGC harbor coamplification of HER2 and/or EGFR concurrently in the same tumor cells, which can drive de novo resistance. One patient with concurrent MET and HER2 amplification was refractory to HER2 blockade, but responded to combined MET/HER2 inhibition. We also found striking heterogeneity in MET amplification between distinct metastatic lesions and primary tumors in individual patients with EGC. In these patients, MET inhibition led to mixed responses and disease progression through outgrowth of non-MET-amplified clones, which could be monitored in circulating tumor DNA. Thus, receptor coamplification and molecular heterogeneity may be key drivers of clinical resistance in MET-amplified EGC. SIGNIFICANCE: Coamplification of driver oncogenes occurs frequently in EGC and can drive therapeutic resistance, supporting a role for comprehensive molecular analysis prior to targeted therapy. EGCs can also exhibit extensive heterogeneity in gene amplification between distinct tumor lesions within the same patient, suggesting that molecular profiling of a single-lesion biopsy may be insufficient to guide targeted therapy selection.


Subject(s)
Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/genetics , Gene Amplification , Genetic Heterogeneity , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/drug therapy , Humans , In Situ Hybridization, Fluorescence , Molecular Targeted Therapy , Mutation , Positron-Emission Tomography , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/drug therapy , Tomography, X-Ray Computed , Treatment Outcome
3.
Cancer Discov ; 5(4): 358-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673644

ABSTRACT

UNLABELLED: BRAF mutations occur in approximately 10% of colorectal cancers. Although RAF inhibitor monotherapy is highly effective in BRAF-mutant melanoma, response rates in BRAF-mutant colorectal cancer are poor. Recent clinical trials of combined RAF/EGFR or RAF/MEK inhibition have produced improved efficacy, but patients ultimately develop resistance. To identify molecular alterations driving clinical acquired resistance, we performed whole-exome sequencing on paired pretreatment and postprogression tumor biopsies from patients with BRAF-mutant colorectal cancer treated with RAF inhibitor combinations. We identified alterations in MAPK pathway genes in resistant tumors not present in matched pretreatment tumors, including KRAS amplification, BRAF amplification, and a MEK1 mutation. These alterations conferred resistance to RAF/EGFR or RAF/MEK combinations through sustained MAPK pathway activity, but an ERK inhibitor could suppress MAPK activity and overcome resistance. Identification of MAPK pathway reactivating alterations upon clinical acquired resistance underscores the MAPK pathway as a critical target in BRAF-mutant colorectal cancer and suggests therapeutic options to overcome resistance. SIGNIFICANCE: RAF inhibitor combinations represent promising approaches in clinical development for BRAF-mutant colorectal cancer. Initial characterization of clinical acquired resistance mechanisms to these regimens identified several MAPK pathway alterations driving resistance by reactivating MAPK signaling, highlighting the critical dependence of BRAF-mutant colorectal cancers on MAPK signaling and offering potential strategies to overcome resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , MAP Kinase Signaling System/drug effects , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Gene Amplification , Humans , MAP Kinase Kinase 1/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Oncogene Protein p21(ras)/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Transcriptional Activation
4.
Cancer Discov ; 4(4): 452-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24444711

ABSTRACT

Although the roles of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling in KRAS-driven tumorigenesis are well established, KRAS activates additional pathways required for tumor maintenance, the inhibition of which are likely to be necessary for effective KRAS-directed therapy. Here, we show that the IκB kinase (IKK)-related kinases Tank-binding kinase-1 (TBK1) and IKKε promote KRAS-driven tumorigenesis by regulating autocrine CCL5 and interleukin (IL)-6 and identify CYT387 as a potent JAK/TBK1/IKKε inhibitor. CYT387 treatment ablates RAS-associated cytokine signaling and impairs Kras-driven murine lung cancer growth. Combined CYT387 treatment and MAPK pathway inhibition induces regression of aggressive murine lung adenocarcinomas driven by Kras mutation and p53 loss. These observations reveal that TBK1/IKKε promote tumor survival by activating CCL5 and IL-6 and identify concurrent inhibition of TBK1/IKKε, Janus-activated kinase (JAK), and MEK signaling as an effective approach to inhibit the actions of oncogenic KRAS.


Subject(s)
Autocrine Communication , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Pyrimidines/pharmacology , Signal Transduction/drug effects , ras Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Chemokine CCL5/metabolism , Human Umbilical Vein Endothelial Cells , Humans , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Mice , Neoplasms, Experimental , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
5.
Sci Transl Med ; 5(196): 196ra98, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23903755

ABSTRACT

RAF and MEK (mitogen-activated or extracellular signal-regulated protein kinase kinase) inhibitors are effective in treating patients with BRAF-mutant melanoma. However, most responses are partial and short-lived, and many patients fail to respond at all. We found that suppression of TORC1 activity in response to RAF or MEK inhibitors, as measured by decreased phosphorylation of ribosomal protein S6 (P-S6), effectively predicted induction of cell death by the inhibitor in BRAF-mutant melanoma cell lines. In resistant melanomas, TORC1 activity was maintained after treatment with RAF or MEK inhibitors, in some cases despite robust suppression of mitogen-activated protein kinase (MAPK) signaling. In in vivo mouse models, suppression of TORC1 after MAPK inhibition was necessary for induction of apoptosis and tumor response. Finally, in paired biopsies obtained from patients with BRAF-mutant melanoma before treatment and after initiation of RAF inhibitor therapy, P-S6 suppression predicted significantly improved progression-free survival. Such a change in P-S6 could be readily monitored in real time by serial fine-needle aspiration biopsies, making quantitation of P-S6 a valuable biomarker to guide treatment in BRAF-mutant melanoma.


Subject(s)
Melanoma/enzymology , Melanoma/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Multiprotein Complexes/antagonists & inhibitors , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Indoles/pharmacology , Indoles/therapeutic use , Mechanistic Target of Rapamycin Complex 1 , Melanoma/drug therapy , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Ribosomal Protein S6/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Vemurafenib , Xenograft Model Antitumor Assays
6.
Cancer Cell ; 23(1): 121-8, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23245996

ABSTRACT

KRAS is the most commonly mutated oncogene, yet no effective targeted therapies exist for KRAS mutant cancers. We developed a pooled shRNA-drug screen strategy to identify genes that, when inhibited, cooperate with MEK inhibitors to effectively treat KRAS mutant cancer cells. The anti-apoptotic BH3 family gene BCL-XL emerged as a top hit through this approach. ABT-263 (navitoclax), a chemical inhibitor that blocks the ability of BCL-XL to bind and inhibit pro-apoptotic proteins, in combination with a MEK inhibitor led to dramatic apoptosis in many KRAS mutant cell lines from different tissue types. This combination caused marked in vivo tumor regressions in KRAS mutant xenografts and in a genetically engineered KRAS-driven lung cancer mouse model, supporting combined BCL-XL/MEK inhibition as a potential therapeutic approach for KRAS mutant cancers.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Drug Screening Assays, Antitumor , Humans , Mice , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...